WO2024062476A1 - Anticorps humanisés dirigés contre la nectine-4 et conjugués médicamenteux associés - Google Patents

Anticorps humanisés dirigés contre la nectine-4 et conjugués médicamenteux associés Download PDF

Info

Publication number
WO2024062476A1
WO2024062476A1 PCT/IL2023/051016 IL2023051016W WO2024062476A1 WO 2024062476 A1 WO2024062476 A1 WO 2024062476A1 IL 2023051016 W IL2023051016 W IL 2023051016W WO 2024062476 A1 WO2024062476 A1 WO 2024062476A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
sequence
antibody
chain
cancer
Prior art date
Application number
PCT/IL2023/051016
Other languages
English (en)
Inventor
Pinchas TSUKERMAN
Anas ATIEH
Akram OBIEDAT
Alon VITENSHTEIN
Guy CINAMON
Original Assignee
Nectin Therapeutics Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nectin Therapeutics Ltd. filed Critical Nectin Therapeutics Ltd.
Publication of WO2024062476A1 publication Critical patent/WO2024062476A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68031Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being an auristatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6861Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from kidney or bladder cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the invention is in the field of immunotherapy and relates to humanized anti-Nectin-4 antibodies and to antibody-conjugates and therapeutic and diagnostic compositions comprising them, for treating diseases, in particular cancer.
  • Cancer immunotherapy is utilized for generating and augmenting an anti-tumor immune response, e.g., by treatment with antibodies specific to antigens on tumor cells, or by specific activation of anti-tumor T cells.
  • the ability of recruiting immune cells (e.g., T cells) against tumor cells in a patient provides a therapeutic modality of fighting cancer types and metastasis that are otherwise considered incurable.
  • Nectin-4 also termed poliovirus receptor-related 4 (PVRL4), is a type I transmembrane protein and member of the Nectin family of related immunoglobulin-like adhesion molecules.
  • Nectin-4 is a tumor associated marker for many tumors including bladder cancer, breast cancer, lung cancer and other malignancies.
  • Nectin-4 Enfortumab antibody-drug conjugate as a highly potent therapeutic agent in multiple preclinical cancer models.
  • the antibody, conjugated with the microtubule inhibitor vedotin binds human, as well as rat and monkey Nectin-4 and inhibits growth of several cell lines and xenografts that express Nectin-4.
  • WO 2019/215728 discloses monoclonal antibodies that recognize human Nectin-4 with high affinity and specificity and inhibit its binding to T cell immunoreceptor with Ig and ITIM domains (TIGIT).
  • ADCs Antibody-drug conjugates
  • mAbs linked to cytotoxic drugs payloads
  • Auristatin is a microtubule-destroying drug. It was derived from marine shell-less mollusk Dolabella auricularia called dolastatins.
  • MMAE monomethyl auristatin E
  • MMAF monomethyl auristatin F
  • MMAE and MMAF were developed by Seattle Genetics and used as payloads for ADCs. MMAF and MMAE have their advantages and disadvantages. MMAE is more membrane- permeable and has a lower IC50 than MMAF. However, MMAF is more hydrophilic and has a lower aggregation tendency to show lower systemic toxicity than MMAE (park et al. Molecules 2019, 24, 2754).
  • the present invention provides humanized antibodies that specifically bind Nectin-4, or antigen binding portion thereof.
  • the humanized antibodies of the present invention selected from a larger collection of antibody clones, have improved properties compared to other anti- Nectin-4 antibodies.
  • the present invention further provides, according to some embodiments, conjugates comprising the antibodies and therapeutic or diagnostic agents.
  • the conjugates comprise a cytotoxic moiety and are useful in treating cancer having tumor cells presenting the Nectin-4 receptor on their surface.
  • NTX1105 ADC is more efficient than the most advanced ADC bearing an anti-Nectin-4 antibody published so far (PADCEV or enfortumab-based ADCs). It is now shown that NTX1105-based ADCs have significant anti- tumor activity in treatment regiments in which PADCEV, or other enfortumab-based ADCs have no activity.
  • a large collection of humanized antibodies was produced by combining specific sets of complementarity determining regions (CDR) sequences and human framework sequences and introducing specific mutations in these sequences to produce antibodies with modified variable regions and improved properties.
  • the antibodies disclosed herein were designed based on factors including homology, T-cell epitopes, key residues, and predicted structures.
  • the newly designed humanized variable regions described herein preserve the residues critical for the maintenance of the antibody’s conformation and binding affinity, while having significantly lower incidence of potential T cell epitopes, thus minimizing the risk of adverse immune response towards the antibodies.
  • the humanized antibodies disclosed herein had superior productivity characteristic, and improved developability properties (e.g higher resistance to aggregation as measured by improved Tagg).
  • the humanized antibodies disclosed herein were found to be highly suitable for use as targeted therapy with therapeutic toxins. It is now disclosed that the anti-Nectin-4 monoclonal humanized antibodies described herein, conjugated to a cytotoxic moiety, exhibit robust killing of various tumor cell lines.
  • the direct targeting of toxins using the antibodies described herein has the potential to increase the anti-tumor activity of these toxins and to improve the survival of cancer patients.
  • Some of the ADCs of the present invention comprise sequence modification of their Fc region that significantly reduces their binding by FcyR- bearing normal cells, thereby increasing their safety.
  • the present invention provides a humanized antibody that specifically binds human Nectin-4, or a fragment thereof comprising at least the antigen binding site, wherein the antibody or a fragment thereof comprises a heavy chain and a light chain, wherein the heavy chain comprises a variable region having an amino acid sequence at least about 90% identical to a sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6; and wherein the light chain comprises a variable region having an amino acid sequence at least about 90% identical to a sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12.
  • the humanized antibody or a fragment thereof comprises a heavy chain comprising a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 2. According to some embodiments, the humanized antibody or a fragment thereof comprises a heavy chain comprising a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 3. According to some embodiments, the humanized antibody or a fragment thereof comprises a heavy chain comprising a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 4. According to some embodiments, the humanized antibody or a fragment thereof comprises a heavy chain comprising a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 5. According to some embodiments, the humanized antibody or a fragment thereof comprises a heavy chain comprising a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 6.
  • the humanized antibody or a fragment thereof comprises a light chain comprising a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 8. According to some embodiments, the humanized antibody or a fragment thereof comprises a light chain comprising a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 9. According to some embodiments, the humanized antibody or a fragment thereof comprises a light chain comprising a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 10. According to some embodiments, the humanized antibody or a fragment thereof comprises a light chain comprising a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 11. According to some embodiments, the humanized antibody or a fragment thereof comprises a light chain comprising a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 12.
  • the humanized antibody or a fragment thereof comprises a heavy chain and a light chain, wherein the heavy chain comprises a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 13, and the light chain comprises a variable region having an amino acid sequence at least about 95% identical to SEQ ID NO: 14.
  • the humanized antibody or a fragment thereof comprises a heavy chain comprising a variable region having the amino acid set forth in SEQ ID NO: 13.
  • the humanized antibody or a fragment thereof comprises a light chain comprising a variable region having the amino acid set forth in SEQ ID NO: 14.
  • the humanized antibody or a fragment thereof comprises a heavy chain and a light chain, wherein the heavy-chain variable region comprises the sequence set forth in SEQ ID NO: 13, and the light-chain variable region comprises the sequence set forth in SEQ ID NO: 14.
  • CDR sequences of a given antibody molecule There are several methods known in the art for determining the CDR sequences of a given antibody molecule, but there is no standard unequivocal method. Determination of CDR sequences from antibody heavy and light chain variable regions can be made according to any method known in the art, including but not limited to the methods known as KAB AT, Chothia and IMGT.
  • a selected set of CDRs may include sequences identified by more than one method, namely, some CDR sequences may be determined using KABAT and some using IMGT, for example.
  • the CDR sequences of the mAb variable regions are determined using the IMGT method.
  • the humanized antibody or a fragment thereof comprises a set of six CDR sequences, wherein the heavy-chain CDR1 comprising the sequence SYY (SEQ ID NO: 26), heavy-chain CDR2 comprising the sequence IYPGNVNT (SEQ ID NO: 22), heavy-chain CDR3 comprising the sequence SNPYVMDY (SEQ ID NO: 17), light-chain CDR1 comprising the sequence QSVNND (SEQ ID NO: 24), light-chain CDR2 comprising the amino acid sequence YAS (SEQ ID NO: 25), and light-chain CDR3 comprising the sequence QQAYRSPYT (SEQ ID NO: 20).
  • the humanized antibody or a fragment thereof comprises a set of six CDR sequences, wherein heavy-chain CDR1 comprising the sequence SYYIH (SEQ ID NO: 15), heavy-chain CDR2 comprising the sequence WIYPGNVNTKYNERF(K/Q)G (SEQ ID NO: 16), heavy-chain CDR3 comprising the sequence SNPYVMDY (SEQ ID NO: 17), light-chain CDR1 comprising the sequence (K/R)ASQSVNNDVA (SEQ ID NO: 18), light-chain CDR2 comprising the sequence YASNRFT (SEQ ID NO: 19), and light-chain CDR3 comprising the sequence QQAYRSPYT (SEQ ID NO: 20).
  • heavy-chain CDR1 comprising the sequence SYYIH (SEQ ID NO: 15
  • heavy-chain CDR2 comprising the sequence WIYPGNVNTKYNERF(K/Q)G (SEQ ID NO: 16
  • heavy-chain CDR3 comprising the sequence SNPYVMDY
  • the humanized antibody or a fragment thereof comprises a heavy-chain CDR2 comprising the sequence WIYPGNVNTKYNERFKG (SEQ ID NO: 27). According to some embodiments, the humanized antibody or a fragment thereof comprises a heavy-chain CDR2 comprising the sequence WIYPGNVNTKYNERFQG (SEQ ID NO: 28). According to some embodiments, the humanized antibody or a fragment thereof comprises a light-chain CDR1 comprising the sequence KASQSVNNDVA (SEQ ID NO: 29). According to some embodiments, the humanized antibody or a fragment thereof comprises a light-chain CDR1 comprising the sequence RASQSVNNDVA (SEQ ID NO: 30).
  • the humanized antibody or a fragment thereof comprises a set of six CDR sequences, wherein heavy-chain CDR1 comprising the sequence GYTFTSYY (SEQ ID NO: 21), heavy-chain CDR2 comprising the sequence IYPGNVNT (SEQ ID NO: 22), heavy-chain CDR3 comprising the sequence ARSNPYVMDY (SEQ ID NO: 23), Light-chain CDR1 comprising the sequence QSVNND (SEQ ID NO: 24), Light- chain CDR2 comprising the amino acid sequence YAS (SEQ ID NO: 25), and Light-chain CDR3 comprising the sequence QQAYRSPYT (SEQ ID NO: 20).
  • the humanized antibody or antigen binding fragment thereof comprising: a heavy chain variable region and a light chain variable region, the heavy chain variable region comprising a set of four heavy chain (HC) framework (FR) sequences: (A) FR-H1 selected from the group consisting of SEQ ID NOs: 31, 32, and 33; (B) FR-H2 selected from the group consisting of SEQ ID NOs: 34, 35, and 36; (C) FR-H3 selected from the group consisting of SEQ ID NOs: 37, 38, 39, and 40; (D) FR-H4 is SEQ ID NO: 41; and the light chain variable region comprising a set of four light chain (LC) framework (FR) sequences: (A) FR-L1 selected from the group consisting of SEQ ID NOs: 42, 43, and 44; (B) FR-L2 selected from the group consisting of SEQ ID NOs: 45 and 46; (C) FR-L3 selected from the group consisting of SEQ ID NO
  • the heavy chain variable region of the humanized monoclonal antibody comprises an amino acid sequence at least about 97% identical to a sequence selected from the group consisting of SEQ ID NOs: 2, 3, 4, 5, and 6; and the light chain variable region of the humanized monoclonal antibody comprises an amino acid sequence at least about 97% identical to a sequence selected from the group consisting of SEQ ID NOs: 8, 9, 10, 11, and 12.
  • the heavy chain variable region of the humanized monoclonal antibody comprises a sequence selected from the group consisting of SEQ ID NOs: 2, 3, 4, 5, and 6; and the light chain variable region of the humanized monoclonal antibody comprises a sequence selected from the group consisting of SEQ ID NOs: 8, 9, 10, 11, and 12.
  • Each combination of heavy and light chain variable region represents a separate embodiment of the invention.
  • the heavy chain variable region of the humanized monoclonal antibody comprises the SEQ ID NO: 5; and the light chain variable region of the humanized monoclonal antibody comprises the SEQ ID NO: 12 (denoted herein NTX1105 (H4/k5)).
  • the heavy chain variable region of the humanized monoclonal antibody comprises the SEQ ID NO: 6; and the light chain variable region of the humanized monoclonal antibody comprises the SEQ ID NO: 12.
  • the humanized antibody or fragment thereof is a monoclonal antibody, Fab, F(ab)2, a single-domain antibody, or a single chain variable fragment (scFv).
  • the humanized antibody or fragment thereof is an IgG monoclonal antibody.
  • the humanized antibody has a heavy chain constant region selected from IgGl, IgG4, and IgG2.
  • the humanized antibody or fragment thereof is an IgG4 subclass.
  • the humanized antibody or antigen binding fragment thereof is an IgGl subclass.
  • the antibody has a kappa light chain constant region.
  • the humanized antibody has a mutated Fc domain that prevents FcyR-mediated internalization.
  • the humanized antibody comprises a Fc null domain.
  • the Fc domain is null for binding to Fc ⁇ receptors found on immune cells.
  • the Fc domain is null for binding to CD64, CD32a, CD32b, CD16a, and/or CD16b.
  • the humanized antibody comprises a Fc null domain having the EAEAPG mutant.
  • the humanized antibody comprising a heavy chain sequence set forth in SEQ ID NO: 51, and a light chain sequence set forth in SEQ ID NO: 52 (NTX1105 (H4/k5) having EAEAPG mutant (FcgR null )).
  • NTX1105 H4/k5 having EAEAPG mutant (FcgR null )
  • a conjugate comprising the humanized antibody or fragment thereof described above is provided.
  • Antibodies or fragments thereof according to the present invention may be attached to a cytotoxic moiety, a radioactive moiety, or a labeling tag.
  • the humanized antibody or fragment thereof is conjugated to a toxin (payload).
  • the toxin is selected from the group consisting of microtubule inhibitor, DNA synthesis inhibitor, topoisomerase inhibitor, and RNA polymerase inhibitor.
  • the toxin is a microtubule-destroying drug.
  • the toxin is auristatin or a derivative thereof.
  • the auristatin derivative is monomethyl auristatin E (MMAE) or monomethyl auristatin F (MMAF).
  • the toxin is saporin.
  • the toxin is a maytansine derivative.
  • the maytansine derivative is DM4 or DM1.
  • the toxin is quinoline alkaloid.
  • the quinoline alkaloid is SN-38.
  • the toxin is directly linked to the antibody.
  • the antibody and the toxin are linked through a linker.
  • the toxin is covalently linked to the humanized antibody directly or through a linker.
  • the linker is cleavable. According to additional embodiments, the linker is not cleavable. According to some embodiments, the linker is an enzymatic cleavable linker. According to certain embodiments, the linker is a pH-sensitive linker. According to some embodiments, the linker is a reducible linker (sulfo-SPDB).
  • the linker is selected from the group consisting of
  • MC Maleimidocaproyl
  • MC-VC-PAB Maleimidocaproyl-Valine-Citrulline- p-amino-benzyloxycarbonyl
  • SMCC Maleimidomethyl cyclohexane- 1 -carboxylate
  • SPDB N-succinimidyl-4-(2- pyridyldithio) butanoate
  • the Drug-to-Antibody Ratio is 4-8. According to certain embodiments, the DAR is 4 (DAR-4). According to certain embodiments, the DAR is 8 (DAR-8).
  • polynucleotide sequences encoding the amino acid sequences of the heavy chain variable region and the light chain variable region as described above are provided.
  • the present invention provides a nucleic acid construct comprising a nucleic acid molecule encoding at least one humanized antibody chain or fragment thereof as described herein.
  • the nucleic acid construct is a plasmid.
  • a plasmid for expressing the humanized antibodies or fragment thereof as described herein comprising nucleic acid molecule encoding the antibody.
  • the present invention provides, according to another aspect, a pharmaceutical composition
  • a pharmaceutical composition comprising the humanized antibody or antigen binding fragment described herein or a conjugate comprising the antibody and a pharmaceutically acceptable excipient, carrier, or diluent.
  • the pharmaceutical composition is for use in treating cancer.
  • Any administration mode may be used to deliver the compositions of the present invention to a subject in need thereof, including parenteral and enteral administration modes.
  • the pharmaceutical composition is formulated for injection or infusion. According to some embodiments, the pharmaceutical composition is formulated for intravenous administration. In certain embodiments, the pharmaceutical composition is formulated for intratumoral administration. According to yet another aspect, the present invention provides a method of treating cancer comprising administering to a subject in need thereof, a therapeutically effective amount of at least one humanized antibody, a fragment thereof or their conjugate as described herein.
  • the cancer comprises a solid tumor.
  • the cancer is selected from the group consisting of prostate cancer, colorectal cancer, bladder cancer, liver cancer, ovarian cancer, endometrial cancer, stomach cancer, thyroid cancer, carcinoid tumor, head and neck cancer, breast cancer, pancreatic cancer, testis cancer, urothelial cancer, cervical cancer, melanoma, lymphoma and lung cancer.
  • the cancer is selected from the group consisting of Pancreatic Ductal Adenocarcinoma, Kidney Renal Clear Cell Carcinoma and Skin Cutaneous Melanoma.
  • the cancer is a hematological cancer.
  • the hematological cancer is selected from leukemia including acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphocytic leukemia (ALL), and chronic lymphocytic leukemia (CLL); lymphoma, including Hodgkin disease, and non-Hodgkin lymphoma; and multiple myeloma.
  • the subject is human.
  • the method of treating cancer comprises administering or performing at least one additional anti-cancer therapy.
  • the additional anti-cancer therapy is surgery, chemotherapy, radiotherapy, or immunotherapy.
  • the method of treating cancer comprises administration of the humanized antibody or conjugate described herein and an additional anti-cancer agent.
  • the additional anti-cancer agent is selected from the group consisting of: immune-modulator, activated lymphocyte cell, kinase inhibitor and chemotherapeutic agent.
  • the anti-cancer agent is selected from the group consisting of: erbitux, cytarabine, fludarabine, fluorouracil, mercaptopurine, methotrexate, thioguanine, gemcitabine, vincristine, vinblastine, vinorelbine, carmustine, lomustine, chlorambucil, cyclophosphamide, cisplatin, carboplatin, ifosfamide, mechlorethamine, melphalan, thiotepa, dacarbazine, bleomycin, dactinomycin, daunorubicin, doxorubicin, idarubicin, mitomycin, mitoxantrone, plicamycin, etoposide, teniposide and any combination thereof.
  • erbitux e.g., erbitux, cytarabine, fludarabine, fluorouracil, mercaptopurine, methot
  • the method of treating cancer involves preventing or reducing formation, growth or spread of metastases in a subject.
  • the present invention provides an antibody-drug conjugate (ADC) comprising a humanized anti-Nectin-4 antibody or antigen binding portion thereof, conjugated to a toxin (payload), the antibody or antigen binding portion thereof comprises a set of six CDR sequences, wherein heavy-chain CDR1 comprising the sequence SYY (SEQ ID NO: 26), heavy-chain CDR2 comprising the sequence IYPGNVNT (SEQ ID NO: 22), heavy-chain CDR3 comprising the sequence SNPYVMDY (SEQ ID NO: 17), light-chain CDR1 comprising the sequence QSVNND (SEQ ID NO: 24), light-chain CDR2 comprising the amino acid sequence YAS (SEQ ID NO: 25), and light-chain CDR3 comprising the sequence QQAYRSPYT (SEQ ID NO: 20).
  • ADC antibody-drug conjugate
  • the antibody-drug conjugate comprises a humanized anti-Nectin-4 antibody or antigen binding portion thereof, comprising a set of six CDR sequences, wherein heavy-chain CDR1 comprising the sequence SYYIH (SEQ ID NO: 15), heavy-chain CDR2 comprising the sequence WIYPGNVNTKYNERF(K/Q)G (SEQ ID NO: 16), heavy-chain CDR3 comprising the sequence SNPYVMDY (SEQ ID NO: 17), light- chain CDR1 comprising the sequence (K/R)ASQSVNNDVA (SEQ ID NO: 18), light-chain CDR2 comprising the sequence YASNRFT (SEQ ID NO: 19), and light-chain CDR3 comprising the sequence QQAYRSPYT (SEQ ID NO: 20).
  • heavy-chain CDR1 comprising the sequence SYYIH (SEQ ID NO: 15
  • heavy-chain CDR2 comprising the sequence WIYPGNVNTKYNERF(K/Q)G (SEQ ID
  • the humanized antibody or a fragment thereof comprises a heavy-chain CDR2 comprising the sequence WIYPGNVNTKYNERFKG (SEQ ID NO: 27). According to some embodiments, the humanized antibody or a fragment thereof comprises a heavy-chain CDR2 comprising the sequence WIYPGNVNTKYNERFQG (SEQ ID NO: 28).
  • the humanized antibody or a fragment thereof comprises a light-chain CDR1 comprising the sequence KASQSVNNDVA (SEQ ID NO: 29). According to some embodiments, the humanized antibody or a fragment thereof comprises a light-chain CDR1 comprising the sequence RASQSVNNDVA (SEQ ID NO: 30).
  • the antibody or a fragment thereof comprises a heavy chain and a light chain, wherein the heavy chain comprises a variable region having an amino acid sequence at least about 90% identical to a sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6; and wherein the light chain comprises a variable region having an amino acid sequence at least about 90% identical to a sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12.
  • the heavy chain variable region of the humanized antibody comprises an amino acid sequence set forth in SEQ ID NO. 13; and the light chain variable region of the humanized antibody comprises an amino acid sequence set forth in SEQ ID NO: 14.
  • the heavy chain variable region of the humanized antibody comprises an amino acid sequence at least about 97% identical to a sequence selected from the group consisting of SEQ ID NOs: 2, 3, 4, 5, and 6; and the light chain variable region of the humanized monoclonal antibody comprises an amino acid sequence at least about 97% identical to a sequence selected from the group consisting of SEQ ID NOs: 8, 9, 10, 11, and 12.
  • the heavy chain variable region of the humanized monoclonal antibody comprises a sequence set forth in SEQ ID NO: 5, and the light chain variable region of the humanized monoclonal antibody comprises a sequence set forth in SEQ ID NO: 12.
  • the heavy chain variable region of the humanized monoclonal antibody comprises a sequence set forth in SEQ ID NO: 6, and the light chain variable region of the humanized monoclonal antibody comprises a sequence set forth in SEQ ID NO: 12.
  • the antibody-drug conjugate comprises a toxin as described hereinabove.
  • the toxin is selected from the group consisting of microtubule inhibitor, DNA synthesis inhibitor, topoisomerase inhibitor, and RNA polymerase inhibitor.
  • the toxin is a microtubule-destroying drug.
  • the toxin is auristatin or a derivative thereof.
  • the auristatin derivative is monomethyl auristatin E (MMAE) or monomethyl auristatin F (MMAF).
  • the toxin is saporin.
  • the toxin is a maytansine derivative.
  • the maytansine derivative is DM4 or DM1.
  • the toxin is a quinoline alkaloid.
  • the quinoline alkaloid is SN-38.
  • the toxin is selected from the group consisting of DM4, MMAE and SN-38. According to certain embodiments, the toxin is DM4 or MMAE.
  • the toxin is a topoisomerase I inhibitor. According to some embodiments, the toxin is a derivative of camptothecin. According to certain embodiments, the toxin is Exatecan.
  • the toxin is directly linked to the antibody.
  • the antibody and the toxin are linked through a linker.
  • the toxin is covalently linked to the humanized antibody directly or through a linker.
  • the linker is cleavable. According to other embodiments, the linker is not cleavable. According to some embodiments, the cleavable linker is selected from the group consisting of an enzymatic cleavable linker, a pH-sensitive linker and a reducible linker. According to some embodiments, the linker is an enzymatic cleavable linker. According to certain embodiments, the linker is a pH-sensitive linker. According to some embodiments, the linker is a reducible linker.
  • MC Maleimidocaproyl
  • MC-VC-PAB Maleimidocaproyl-Valine-Citrulline- p-amino-benzyloxycarbonyl
  • SMCC Maleimidomethyl cyclohexane- 1 -carboxylate
  • sulfo-SPDB N-succinimidyl-4-(
  • the conjugate comprises the toxin MMAE and the linker MC-VC-PAB (denoted herein NTX1105-MMAE). According to some embodiments, the conjugate comprises the toxin MMAF and the linker MC (denoted herein NTX1105- MMAF). According to some embodiments, the conjugate comprises the toxin DM1 and the linker SMCC (denoted herein NTX1105-DM1). According to some embodiments, the conjugate comprises the toxin DM4 and the linker SPDB (denoted herein NTX1105-DM4). According to some embodiments, the conjugate comprises the toxin SN38 and the linker Lys- PAB-CO (denoted herein NTX1105-SN38).
  • the ADC comprises an anti-Nectin-4 antibody that competes with an antibody described herein to specifically bind to the Nectin-4 molecule.
  • the ADC comprises an anti-Nectin-4 antibody that competes with an antibody comprising heavy-chain and light chain variable regions wherein heavy-chain CDR1 comprising the sequence SYYIH (SEQ ID NO: 15), heavy-chain CDR2 comprising the sequence WIYPGNVNTKYNERF(K/Q)G (SEQ ID NO: 16), heavy-chain CDR3 comprising the sequence SNPYVMDY (SEQ ID NO: 17), light-chain CDR1 comprising the sequence (K/R)ASQSVNNDVA (SEQ ID NO: 18), light-chain CDR2 comprising the sequence YASNRFT (SEQ ID NO: 19), and light-chain CDR3 comprising the sequence QQAYRSPYT (SEQ ID NO: 20), to specifically bind to the Nectin-4 molecule.
  • heavy-chain CDR1 comprising the sequence
  • the present invention provides, according to another aspect, a pharmaceutical composition
  • a pharmaceutical composition comprising the antibody-drug conjugate described herein and a pharmaceutically acceptable excipient, carrier, or diluent.
  • Any administration mode may be used to deliver the compositions of the present invention to a subject in need thereof, including parenteral and enteral administration modes.
  • the pharmaceutical composition is formulated for injection or infusion. According to some embodiments, the pharmaceutical composition is formulated for intravenous (IV) administration. In certain embodiments, the pharmaceutical composition is formulated for intratumoral (IT) administration. According to some embodiments, the conjugate or the pharmaceutical composition is for use in treating a cancer in an individual.
  • the cancer is as described hereinabove.
  • the cancer comprises a solid tumor.
  • the cancer is selected from the group consisting of liver cancer, lung cancer, colon cancer, glioblastoma, adrenal cancer, uterine cancer, testis cancer, head and neck cancer, pancreatic cancer, and breast cancer. Each possibility represents a separate embodiment of the invention.
  • the use further comprises the use in a combination with an additional ADC.
  • the present invention provides, according to another aspect, a method of treating a cancer in an individual in need of such treatment, the method comprising administering to the individual a therapeutically effective amount of the conjugate or the pharmaceutical composition described herein.
  • the cancer is a solid tumor.
  • the cancer is a non-solid tumor.
  • the cancer is selected from the group consisting of glioblastoma, pancreatic cancer, breast cancer, bladder cancer, kidney cancer, head and neck cancer, ovarian cancer, colon cancer, cervical cancer, prostate cancer, and lung cancer.
  • the method of treating cancer involves preventing or reducing formation, growth or spread of metastases in a subject.
  • the cancer is a hematological cancer.
  • the hematological cancer is selected from leukemia including acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphocytic leukemia (ALL), and chronic lymphocytic leukemia (CLL); lymphoma, including Hodgkin disease, and non-Hodgkin lymphoma; and multiple myeloma.
  • the individual is human.
  • the method of treating cancer comprises administering or performing at least one additional anti-cancer therapy.
  • the additional anticancer therapy is surgery, chemotherapy, radiotherapy, or immunotherapy.
  • the method of treating cancer comprises administration of the conjugate described herein and an additional anti-cancer agent.
  • the additional anti-cancer agent is selected from the group consisting of: immune-modulator, activated lymphocyte cell, kinase inhibitor and chemotherapeutic agent.
  • compositions for treating a cancer in an individual afflicted with cancer comprising admixing the ADCs described herein and a pharmaceutically acceptable excipient, carrier, or diluent.
  • the present invention provides a method of delivering at least one humanized antibody, a fragment thereof or conjugate as described herein to a cell comprising contacting the cell with the humanized antibody, a fragment thereof or conjugate.
  • the method comprises administering the humanized antibody, a fragment thereof or conjugate to cells of a subject.
  • the subject is a human subject.
  • the present invention provides a method of delivering at least one humanized antibody, a fragment thereof or their conjugate as described herein to a cell of a subject, comprising administering the at least one humanized antibody, a fragment thereof or conjugate to the subject.
  • the cell is a tumor cell.
  • the present invention further provides, according to an aspect, a method of diagnosing or prognosing cancer in a subject, the method comprises determining the expression level of Nectin-4 in a biological sample of said subject using at least one antibody conjugate as described herein.
  • the method comprising detecting the antibody or antibody fragment that is bound, and determining the levels of expression of Nectin-4 in the sample.
  • the method comprises comparing the levels of expression to control.
  • the control is a predefined value.
  • the control is a corresponding non-cancerous tissue.
  • the comparison indicates or suggests if the subject has cancer.
  • the method is used for diagnosis of cancer subtype. According to some embodiments, the method is used for determine patient eligibility to receive an anti-cancer therapy. According to certain embodiments, the anti-cancer therapy is an antibody-drug conjugate. According to some embodiments, the method is used for determine patient eligibility to receive a therapy with an ADC as described herein.
  • the present invention further provides, according to another aspect, a method of diagnosing, determining or quantifying the expression of Nectin-4, the method comprising contacting a biological sample with an antibody conjugate as described herein, and measuring the level of complex formation.
  • the method for detecting or quantifying the expression of Nectin-4 comprises the steps of: i. incubating a sample with the antibody conjugate described herein; ii. detecting the bound Nectin-4 using said conjugate.
  • the method further comprises the steps of: iii. comparing the amount of step (ii) to a standard curve obtained from a reference sample containing a known amount of Nectin-4; and iv. calculating the amount of the Nectin-4 in the sample from the standard curve.
  • the sample is a body fluid or solid tissue.
  • the method is performed in-vitro or ex-vivo.
  • a kit for measuring the expression of Nectin-4 in a biological sample comprising at least one conjugate as described herein and means for measuring Nectin-4 expression.
  • the kit further comprising instruction material directing the use of the kit.
  • Figures 1A-1C show the correlation of Nectin-4 expression mRNA levels (high or low as indicated) with survival probability of: Pancreatic Ductal Adenocarcinoma ( Figure 1A), Kidney Renal Clear Cell Carcinoma ( Figure IB) and Skin Cutaneous Melanoma. (Figure 1C) patients. Data sets were obtained from the TCGA site and analyzed using oncolnc.org site (https://doi.org/10.7717/peerj-cs.67). N depicts number of patients included at the analysis.
  • Figure 2 shows a graph showing percent of tumors positive for Nectin-4 expression. Data obtained from proteinatlas.com using the HPA010775 mAb (anti Nectin-4 Sigma-Aldrich®). In 13/20 indications moderate-high membranous expression of Nectin-4 is seen.
  • Figure 3 depicts a dose dependent killing of MDA-MB-468 cell line (breast adenocarcinoma cells) in-vitro using the various linker-payload combinations of NTX1105 (VHO/VKO).
  • Figure 4 depicts in-vivo efficacy of the NTX1105 (VHO/VKO) ADCS against the Triple- negative breast cancer (TNBC) tumor cells line MDA-MB-468 that were implanted subcutaneous (s.c.) to NOD-SCID female mice.
  • TNBC Triple- negative breast cancer
  • Figure 5 demonstrates improved characteristics for some of the humanized variants.
  • the productivity of the parental/chimeric (VHO/VKO) antibody and six purified lead humanized variants was evaluated.
  • Figures 6A-6B show robust in-vivo activity of the humanized variants.
  • the results are shown as tumor volume ( Figure 6A) and as a fold change of the tumor volume along time ( Figure 6B).
  • Figures 7A-7B show differential in-vitro activity of NTX1105 (H4K5) ADC compared to Enfortumab-vedotin (PADCEV). Both ADCs were tested for on-cell binding ( Figure 7A) and in-vitro killing activity (Figure 7B).
  • Figures 8A-8B show that the anti-tumor activity in-vivo of NTX1105 (H4K5) conjugated to MC-VC-PAB-MMAE, at DAR-4 is superior to Enfortumab-vedotin (PADCEV).
  • PADCEV Enfortumab-vedotin
  • Figure 9 depicts in-vivo efficacy of humanized NTX1105(H4K5-FcgR null ) ADCs with either DAR-4 MMAE or DAR-8 Exatecan payload against H322M (lung cancer model) s.c. tumor model in Nude female mice.
  • Figure 10 depicts in-vivo superiority of humanized NTX1105(H4K5-FcgR null ) anti-Nectin-4 ADCs with DAR-4 Exatecan compared to Enfortumab-DAR-4 Exatecan against large (>450mm 3 ) H322M. tumor model in Nude female mice.
  • the present invention provides humanized anti-Nectin-4 antibodies and antibody-drug conjugates, or ADCs, comprising them, which are useful in treating cancer.
  • the ADCs described herein comprise antibodies that are almost fully humanized, thus avoiding the risk of adverse immune response towards the antibodies and are therefore likely to be safe for use in humans.
  • the ADCs described herein were found to be highly potent and better than other known anti-Nectin-4 ADCs.
  • Nectin-4" or “Nectin Cell Adhesion Molecule 4”, as used herein refers to a single-pass type I membrane protein of 510 amino acids and a molecular mass of 55454 Da, also known as PVRL4; LNIR; PRR4; and EDSS1.
  • the Nectin-4 protein contains two immunoglobulin-like (Ig-like) C2-type domains and one Ig-like V-type domain. It is involved in cell adhesion through trans-homophilic and -heterophilic interactions.
  • the soluble form is produced by proteolytic cleavage at the cell surface by the metalloproteinase ADAM17/TACE and the secreted form is found in both breast tumor cell lines and breast tumor patients.
  • An exemplary Nectin-4 according to the invention is set forth in SwissPort, UniPort and GenBank symbols or accession numbers: Q96NY8-NECT4_HUMAN; Q96NY8; B4DQW3; Q96K15; Q96NY8-1; Q96NY8-2; ENSP00000356991; NP_112178.2; XP_005245565.1;
  • the present invention provides a humanized antibody that specifically binds human Nectin-4, or a fragment thereof comprising at least the antigen binding site, wherein the antibody or a fragment thereof comprises a heavy chain and a light chain, wherein the heavy chain comprises a variable region having an amino acid sequence at least about 90% identical to a sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6; and wherein the light chain comprises a variable region having an amino acid sequence at least about 90% identical to a sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12.
  • the present invention provides an antibody-drug conjugate (ADC) comprising a humanized anti-Nectin-4 antibody or antigen binding portion thereof, conjugated to a toxin (payload), the antibody or antigen binding portion thereof comprises a set of six CDR sequences, wherein heavy-chain CDR1 comprising the sequence SYY (SEQ ID NO: 26), heavy-chain CDR2 comprising the sequence IYPGNVNT (SEQ ID NO: 22), heavy-chain CDR3 comprising the sequence SNPYVMDY (SEQ ID NO: 17), Light-chain CDR1 comprising the sequence QSVNND (SEQ ID NO: 24), Light-chain CDR2 comprising the amino acid sequence YAS (SEQ ID NO: 25), and Light-chain CDR3 comprising the sequence QQAYRSPYT (SEQ ID NO: 20).
  • ADC antibody-drug conjugate
  • the present invention comprises an antibody-drug conjugate (ADC) comprising a humanized anti-Nectin-4 antibody, or antigen binding portion thereof, conjugated to a toxin selected from the group consisting of MMAE, SN-38, DM1, DM4, and MMAFA, the humanized antibody or antigen binding portion thereof comprising a heavy chain and a light chain, wherein the heavy chain comprises a variable region having an amino acid sequence at least about 90% identical to a sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6; and wherein the light chain comprises a variable region having an amino acid sequence at least about 90% identical to a sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12.
  • ADC antibody-drug conjugate
  • the humanized antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence QVQL(Q/V)QSG(P/S)ELKKP(E/G)ASVK(I/V)SCKASGYTFTSYYIHWV(K/R)QAPGQG LEW(I/M)GWIYPGNVNTKYNERF(K/Q)GR(A/V)T(L/I)TADKST(N/S)TA(H/Y)MELSSL (T/R)SED(S/T)AVY(F/Y)CARSNPYVMDYWGQGTSVTVSS (SEQ ID NO: 13); and a light chain variable region comprising the amino acid sequence
  • the humanized antibody or a fragment thereof comprises a heavy chain and a light chain, wherein the heavy chain comprises a variable region having an amino acid sequence at least about 95% identical to a sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6; and wherein the light chain comprises a variable region having an amino acid sequence at least about 95% identical to a sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12.
  • SEQ ID NOs: 2-6, 8-12 are humanized variants of NTX1105 that were selected based on improved developability and low immunogenicity.
  • NTX1105 includes the chimeric antibody and/or its humanized variants according to the context.
  • the parent chimeric antibody is disclosed in WO 2019/215728 (clone 11).
  • the parent chimeric antibody is named herein NTX1105 (VHO/VkO).
  • the humanized variants of NTX1105 include Five heavy chain (VH1 to VH5) and five light chains (VKI to VK5). According to some embodiments, the humanized variants comprise the sequence formula set forth in SEQ ID NO: 13 (heavy-chain) and SEQ ID NO: 14 (light-chain).
  • the humanized antibody or a fragment thereof comprises a heavy chain and a light chain, wherein the heavy chain comprises variable region having an amino acid sequence SEQ ID NO: 5 the light chain comprises variable region having an amino acid sequence SEQ ID NO: 12 (denoted herein NTX1105 (VH5/Vk5).
  • the humanized antibody comprises a combination of a heavy chain variable region and a light chain variable region, wherein the combination is selected from the group consisting of: SEQ ID NO: 2 and SEQ ID NO: 8, SEQ ID NO: 2 and SEQ ID NO: 9, SEQ ID NO: 2 and SEQ ID NO: 10, SEQ ID NO: 2 and SEQ ID NO: 11, SEQ ID NO: 2 and SEQ ID NO: 12, SEQ ID NO: 3 and SEQ ID NO: 8, SEQ ID NO: 3 and SEQ ID NO: 9, SEQ ID NO: 3 and SEQ ID NO: 10, SEQ ID NO: 3 and SEQ ID NO: 11, SEQ ID NO: 3 and SEQ ID NO: 12, SEQ ID NO: 4 and SEQ ID NO: 8, SEQ ID NO: 4 and SEQ ID NO: 9, SEQ ID NO: 4 and SEQ ID NO: 10, SEQ ID NO: 4 and SEQ ID NO: 11, SEQ ID NO: 4 and SEQ ID NO: 12, SEQ ID NO: 5 and SEQ ID NO: 8, SEQ ID NO:
  • the humanized antibody comprises a combination of a heavy chain variable region and a light chain variable region, wherein the combination is selected from the group consisting of: i. a heavy chain variable region sequence set forth in SEQ ID NO: 5 and a light chain variable region sequence set forth in SEQ ID NO: 12; and ii. a heavy chain variable region sequence set forth in SEQ ID NO: 6 and a light chain variable region sequence set forth in SEQ ID NO: 12.
  • the conjugates of the invention comprise a humanized antibody as described herein.
  • the antibodies include monoclonal antibodies, polyclonal antibodies, multispecific antibodies (for example, bispecific antibodies and polyreactive antibodies), and antibody fragments.
  • an antibody includes, but is not limited to, full-length, as well as fragments and portion thereof retaining the binding specificities thereof, such as any specific binding portion thereof including those having any number of, immunoglobulin classes and/or isotypes (e.g., IgGl, IgG2, IgG3, IgG4, IgM, IgA, IgD, IgE and IgM); and biologically relevant (antigen-binding) fragments or specific binding portions thereof, including but not limited to Fab, F(ab')2, Fv, and scFv (single chain or related entity).
  • a monoclonal antibody is generally one within a composition of substantially homogeneous antibodies; thus, any individual antibodies comprised within the monoclonal antibody composition are identical except for possible naturally occurring mutations that may be present in minor amounts.
  • the antibody can comprise a human IgGl constant region.
  • the antibody can comprise a human IgG4 constant region.
  • the antibody can comprise a human kappa light chain.
  • antibody herein is used in the broadest sense and includes polyclonal and monoclonal antibodies, including intact antibodies and functional (antigen-binding) antibody fragments thereof, including fragment antigen binding (Fab) fragments, F(ab')2 fragments, Fab' fragments, Fv fragments, recombinant IgG (rlgG) fragments, single chain antibody fragments, including single chain variable fragments (sFv or scFv), and single domain antibodies (e.g., sdAb, sdFv, nanobody) fragments.
  • Fab fragment antigen binding
  • F(ab')2 fragments fragment antigen binding
  • Fab' fragments fragment antigen binding
  • Fv fragments fragment antigen binding
  • rlgG fragment antigen binding fragments
  • single chain antibody fragments including single chain variable fragments (sFv or scFv) fragments.
  • single domain antibodies e.g., sdAb, sdFv, nanobody
  • the term encompasses genetically engineered and/or otherwise modified forms of immunoglobulins, such as intrabodies, peptibodies, fully human antibodies, humanized antibodies, and heteroconjugate antibodies, multispecific, e.g., bispecific, antibodies, diabodies, triabodies, and tetrabodies, tandem di-scFv, tandem tri-scFv.
  • antibody should be understood to encompass functional antibody fragments thereof.
  • the term also encompasses intact or full- length antibodies, including antibodies of any class or sub-class, including IgG and sub-classes thereof, IgM, IgE, IgA, and IgD.
  • the antibody can comprise a human IgGl constant region.
  • the antibody can comprise a human IgG4 constant region.
  • the antibody can comprise a human kappa light chain.
  • CDR sequences of a given antibody molecule There are several methods known in the art for determining the CDR sequences of a given antibody molecule, but there is no standard unequivocal method. Determination of CDR sequences from antibody heavy and light chain variable regions can be made according to any method known in the art, including but not limited to the methods known as KAB AT, Chothia and IMGT.
  • a selected set of CDRs may include sequences identified by more than one method, namely, some CDR sequences may be determined using KABAT and some using IMGT, for example.
  • the CDR sequences of the mAb variable regions are determined using the IMGT method.
  • CDR determination is made according to the Kabat (Wu T.T and Kabat E.A., J Exp Med, 1970; 132:211-50) and IMGT (Lefranc M- P, et al., Dev Comp Immunol, 2003, 27:55-77).
  • antibody fragments refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include, but are not limited to, Fv, Fab, Fab', Fab’-SH, F(ab')2; diabodies; linear antibodies; single- chain antibody molecules (e.g., scFv or sFv); and multispecific antibodies formed from antibody fragments.
  • the antibodies are single-chain antibody fragments comprising a variable heavy chain region and/or a variable light chain region, such as scFvs.
  • a “humanized” antibody is an antibody in which all or substantially all CDR amino acid residues are derived from non-human CDRs and all or substantially all framework region (FR) amino acid residues are derived from human FRs.
  • a humanized antibody optionally may include at least a portion of an antibody constant region derived from a human antibody.
  • a “humanized form” of a non-human antibody refers to a variant of the non-human antibody that has undergone humanization, typically to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the CDR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • a non-human antibody e.g., the antibody from which the CDR residues are derived
  • the amino acid residues in the Fc domain can be substituted to be null, meaning the Fc domain does not bind Fc receptors or can bind with such low affinity and/or avidity as to not cause any Fc receptor signaling as a result of binding.
  • the Fc domain can be null for binding to Fc ⁇ receptors.
  • Some example Fc ⁇ receptors for which the Fc domain can be null for binding can be, but not limited to, FcyRI (CD64), FcyRIIA (CD32a), FcyRIIB (CD32b), FcyRIIIA (CD16a), FcyRIIIA (CD16a) F158 variant, FcyRIIIA (CD16a) V158 variant, or FcyRIIIB (CD 16b).
  • the Fc domain may have one or more, two or more, three or more, or four or more amino acid substitutions that decrease binding of the Fc domain to an Fc receptor.
  • the humanized antibody has a mutated Fc domain that prevents FcyR-mediated internalization.
  • the humanized antibody comprises a Fc null domain.
  • the Fc domain is null for binding to a Fc ⁇ receptors.
  • an "Fc null” refers to a domain that exhibits weak to no binding to one or more of the Fc ⁇ receptors.
  • the humanized antibody comprises a Fc null domain.
  • the Fc domain is null for binding to Fc ⁇ receptors found on immune cells.
  • the Fc domain is null for binding to CD64, CD32a, CD32b, CD16a, and/or CD16b.
  • the humanized antibody comprises a Fc null domain having the LALAPG mutant.
  • the humanized antibody comprising a heavy chain sequence set forth in SEQ ID NO: 51, and a light chain sequence set forth in SEQ ID NO: 52 (NTX1105 (H4/k5) having LALAPG mutant (FcgR null )).
  • the present invention provides conjugates comprising the humanized antibody disclosed herein and a toxin.
  • the toxin is selected from the group consisting of microtubule inhibitor, DNA synthesis inhibitor, topoisomerase inhibitor, and RNA polymerase inhibitor.
  • the toxin is selected from the group consisting of microtubule inhibitor, DNA synthesis inhibitor, topoisomerase inhibitor, and RNA polymerase inhibitor.
  • the toxin is a microtubule-destroying drug.
  • the toxin is auristatin or a derivative thereof.
  • the auristatin derivative is monomethyl auristatin E (MMAE) or monomethyl auristatin F (MMAF).
  • the toxin is Exatecan.
  • the toxin is saponin.
  • the toxin is a maytansine derivative.
  • the maytansine derivative is DM4 or DM1.
  • the toxin is quinoline alkaloid.
  • the quinoline alkaloid is SN-38.
  • the toxin is selected from the group consisting of MMAE, MMAF, Saporin, DM4, DM1, SN-38, Calicheamicin, DXd, PBD, Duocarmycin, Sandramycin, alpha- Amanitin, Chaetocin, CYT997, Daunorubicin, 17-AAG, Agrochelin A, Doxorubicin, Methotrexate, Colchicine, Cordycepin, Epothilone B, Hygrolidin, Herboxidiene, Ferulenol, Curvulin, paclitaxel, Englerin A, Taltobulin, Triptolide, Cryptophycin, and Nemorubicin.
  • MMAE MMAE
  • MMAF Saporin
  • DM4 DM1, SN-38
  • Calicheamicin DXd
  • PBD Duocarmycin
  • Sandramycin alpha- Amanitin
  • Chaetocin Chaetocin
  • CYT997 Daun
  • the toxin is SN-38. According to some embodiments, the toxin is DM1. According to some embodiments, the toxin is DM4. According to some embodiments, the toxin is MMAE. According to some embodiments, the toxin is MMAF. According to some embodiments, the toxin is Exatecan.
  • the antibody is directly linked to the toxin.
  • the antibody and the toxin are linked through a linker.
  • the humanized described herein is covalently linked to the toxin.
  • the linker is cleavable. According to additional embodiments, the linker is not cleavable.
  • the linker is cleaved in response to changes in pH or redox potential. According to some embodiments, the linker is cleaved when contacted with lysosomal enzymes.
  • the linker comprises a portion which is selected from the group consisting of 6-maleimidocaproyl (MC), maleimidopropionyl (MP), valine-citrulline (val-cit), alanine-phenylalanine (ala-phe), p-aminobenzyloxycarbonyl (PAB), N-succinimidyl 4-(2-pyridylthio)valerate (SPP), N-succinimidyl 4-(N-maleimidomethyl)-cyclohexane-l- carboxylate (SMCC), N-succinimidyl (4-iodo-acetyl) aminobenzoate (SLAB), 6- maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl (MC-vc-PAB), Val-Cit- PABC, N-succinimidyl-4-(2-pyridyldithio)butano
  • the antibody is conjugated to two or more molecules of toxin.
  • the present invention provides, according to another aspect, a pharmaceutical composition comprising the conjugate described herein and a pharmaceutically acceptable excipient, carrier, or diluent.
  • the pharmaceutical composition according to the invention is for use in treating cancer.
  • the cancer amendable for treatment by the present invention includes, but is not limited to: carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer, lung cancer (including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung), cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer (including gastrointestinal cancer), pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancer, as well as B-cell lymphoma (including low grade/follicular non- Hodgkin's lymphoma
  • the cancer is selected from the group consisting of breast cancer, colorectal cancer, rectal cancer, non-small cell lung cancer, non-Hodgkins lymphoma (NHL), renal cell cancer, prostate cancer, liver cancer, pancreatic cancer, soft-tissue sarcoma, Kaposi's sarcoma, carcinoid carcinoma, head and neck cancer, melanoma, ovarian cancer, mesothelioma, and multiple myeloma.
  • the cancerous conditions amendable for treatment of the invention include metastatic cancers.
  • the pharmaceutical composition according to the invention are for use in treating cancer characterized by overexpression of Nectin-4.
  • Nectin-4 overexpression related cancer types can be identified using known data bases such as The Cancer Genome Atlas (TCGA).
  • the cancer treatable with a composition according to the present invention is selected from the group consisting of adrenocortical carcinoma (ACC), chromophobe renal cell carcinoma (KICH), liver hepatocellular carcinoma (LIHC), colon and rectal adenocarcinoma (COAD and READ), pancreatic ductal adenocarcinoma (PAAD), pheochromocytoma & paraganglioma (PCPG), papillary kidney carcinoma (KIRP), lung adenocarcinoma (LU AD), head and neck squamous cell carcinoma (HNSC), prostate adenocarcinoma (PRAD), uterine corpus endometrial carcinoma (UCEC), cervical cancer (CESC), cutaneous mela
  • ACC ad
  • the molecules of the present invention as active ingredients are dissolved, dispersed or admixed in an excipient that is pharmaceutically acceptable and compatible with the active ingredient as is well known.
  • excipients are, for example, water, saline, phosphate buffered saline (PBS), dextrose, glycerol, ethanol, or the like and combinations thereof.
  • PBS phosphate buffered saline
  • dextrose glycerol
  • ethanol ethanol
  • suitable carriers are well known to those skilled in the art.
  • the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents.
  • the pharmaceutical composition according to the present invention may be administered together with an anti-neoplastic composition.
  • treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented. The term “treatment” further relates to alleviation or prevention of symptoms associated with the disease, and/or reducing the severity of the disease.
  • cancer refers to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • examples of cancer include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More particular examples of such cancers include melanoma, lung, thyroid, breast, colon, prostate, hepatic, bladder, renal, cervical, pancreatic, leukemia, lymphoma, myeloid, ovarian, uterus, sarcoma, biliary, or endometrial cancer.
  • the method of treating cancer comprises administering the pharmaceutical composition as part of a treatment regimen comprising administration of at least one additional anti-cancer agent.
  • the term “individual,” “patient,” or “subject” refers to individuals diagnosed with, suspected of being afflicted with, or at-risk of developing at least one disease for which the described compositions and method are useful for treating.
  • the individual is a mammal.
  • the mammal is a mouse, rat, rabbit, dog, cat, horse, cow, sheep, pig, goat, llama, alpaca, or yak.
  • the individual is a human.
  • an “effective amount” refers to the amount of a therapeutic that causes a biological effect when administered to a mammal. Biological effects include, but are not limited to, reduced tumor growth, reduced tumor metastasis, or prolonged survival of an animal bearing a tumor.
  • a “therapeutic amount” is the concentration of a drug calculated to exert a therapeutic effect.
  • a therapeutic amount encompasses the range of dosages capable of inducing a therapeutic response in a population of individuals.
  • the mammal can be a human individual.
  • the human individual can be afflicted with or suspected or being afflicted with a tumor.
  • the molecules of the present invention as active ingredients are dissolved, dispersed or admixed in an excipient that is pharmaceutically acceptable and compatible with the active ingredient as is well known.
  • excipients are, for example, water, saline, phosphate buffered saline (PBS), dextrose, glycerol, ethanol, or the like and combinations thereof.
  • PBS phosphate buffered saline
  • dextrose glycerol
  • ethanol ethanol
  • suitable carriers are well known to those skilled in the art.
  • the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents.
  • the method of treating cancer comprises administering the pharmaceutical composition as part of a treatment regimen comprising administration of at least one additional anti-cancer agent.
  • the anti-cancer agent is selected from the group consisting of an antimetabolite, a mitotic inhibitor, a taxane, a topoisomerase inhibitor, a topoisomerase II inhibitor, an asparaginase, an alkylating agent, an antitumor antibiotic, and combinations thereof. Each possibility represents a separate embodiment of the invention.
  • the antimetabolite is selected from the group consisting of cytarabine, fludarabine, fluorouracil, mercaptopurine, methotrexate, thioguanine, gemcitabine, and hydroxyurea.
  • the mitotic inhibitor is selected from the group consisting of vincristine, vinblastine, and vinorelbine.
  • the topoisomerase inhibitor is selected from the group consisting of topotecan and irinotecan.
  • the alkylating agent is selected from the group consisting of busulfan, carmustine, lomustine, chlorambucil, cyclophosphamide, cisplatin, carboplatin, ifosfamide, mechlorethamine, melphalan, thiotepa, dacarbazine, and procarbazine.
  • the antitumor antibiotic is selected from the group consisting of bleomycin, dactinomycin, daunorubicin, doxorubicin, idarubicin, mitomycin, mitoxantrone, and plicamycin.
  • the topoisomerase II is selected from the group consisting of etoposide and teniposide. Each possibility represents a separate embodiment of the present invention.
  • compositions for treating a cancer in an individual afflicted with cancer comprising admixing the humanized antibody or the ADC as described herein and a pharmaceutically acceptable excipient, carrier, or diluent.
  • the cancer comprises a solid tumor.
  • the cancer is selected from the group consisting of glioblastoma, colon cancer, pancreatic cancer, breast cancer, bladder cancer, kidney cancer, head and neck cancer, ovarian cancer, cervical cancer, prostate cancer, and lung cancer.
  • Example 1 High expression of Nectin-4 mRNA correlates with poor survival probability of various cancer patients.
  • Nectin-4 mRNA expression and survival probability was examined on data from TCGA site, analyzed using the oncolnc.org site (https:// doi.org/10.7717/peerj-cs.67) and presented in Figures 1A-1C.
  • the Nectin-4 mRNA expression levels were the basis for dividing patients for two subgroups of low and high expressors, as indicated by the boxes, the survival advantage of low-expressing patients confirms the detrimental role Nectin-4 plays, and the potential to improve survival by targeting this protein.
  • Nectin-4 is expressed at the protein level on the majority of solid tumors
  • the database Proteinatlas.com was searched for all the aliases of Nectin-4 (NECTIN4, PVRL4). Under the pathology rubric, data using a single mAb was found (HPA010775). The expression data across different tumors is depicted in Figure 2. The graph is showing percent of tumors positive for Nectin-4 expression. In all indications at least one patient has surface expression of Nectin-4 and in 13/20 indications membranous expression of Nectin-4 is seen at Moderate-High levels.
  • Example 3 Various cytotoxic agents can be efficiently conjugate to Nectin-4 targeting mAb for the generation of Nectin-4 ADC
  • DAR drug-antibody -ratio
  • Various cytotoxic agents can induce tumor cells killing when conjugated to Nectin-4 targeting mAb in-vitro
  • a dose dependent killing activity of the various linker-payload combinations of NTX1105 was examined.
  • In vitro assay of TNBC model MDA-MB-468 cell line was used. The target cells were plated at 2*10 3 cells per well and allowed to adhere over 4-6 hours period. The ADCs were added at concentrations of 30-1.9 nM, using 2-fold dilutions, and then the cells were incubated with the ADCs for additional 72 hours. After 72 hours, the assay was harvested and tumor cell killing was evaluated using CellTiter-Glo® 2.0 Cell Viability Assay - (Promega G9242) following standard protocol.
  • Example 5 Various cytotoxic agents can induce tumor cells killing when conjugated to Nectin-4 targeting mAb in-vivo
  • Example 6 Binding of human Nectin-4 and cross-reactivity to Cyno Nectin-4 is preserved for the top selected humanized Nectin-4 mAbs
  • Single cycle kinetic data was obtained using recombinant human (Aero Biosystems, Newark, USA) or cynomolgus Nectin-4 (Aero Biosystems, Newark, USA) as the analyte injected at a flow rate of 40 pl/min to minimize any potential mass transfer effects.
  • a four point, two-fold dilution range from 1.25 nM to 10 nM of antigen in running buffer was used without regeneration between each concentration.
  • the association phases were monitored for 150 seconds for each of the four injections of increasing concentrations of antigen and a single dissociation phase was measured for 250 seconds following the last injection of antigen (in order to improve fit the dissociation was cropped to 175 seconds for human Nectin-4).
  • Example 7 Improved characteristics of the humanized anti-Nectin-4 mAbs (Reduced immunogenicity)
  • the designed VH and VK sequences were analyzed for the occurrence of potential T cell epitopes as determined by application of Abzena’s proprietary in silico technology, iTopeTM (Perry et al. 2008).
  • the iTopeTM software predicts favorable interactions between amino acid side chains of a peptide and specific binding pockets (in particular pocket positions; pl, p4, p6, p7 and p9) within the open-ended binding grooves of 34 human MHC class II alleles. These alleles represent the most common HLA-DR alleles found world-wide with no weighting attributed to those found most prevalently in any particular ethnic population.
  • Promiscuous high affinity MHC Class II binding peptides bind > 50% of alleles with a majority (17 out of 34 alleles) having a binding score > 0.6.
  • a promiscuous peptide is defined as binding to 10 or more of the subset of 20 alleles.
  • a number of germline promiscuous high and moderate affinity MHC Class II binding ligands were identified in the parental antibody and designed variants however it is unlikely that these epitopes have immunogenic potential due to T cell tolerance, and so were excluded from any further analysis. Overall, the process of humanization resulted in significantly improved mAbs, having lower immunogenicity score, and thus lower associated risk.
  • the least immunogenic variants after this step included heavy chains variants 3-5 and light chain 5 (Table 3).
  • Table 3 Improved characteristics of the humanized anti-Nectin-4 mAbs. Reduced immunogenicity as measured by predicted MHCII epitopes (less predicted epitopes correlates to lower immunogenicity) (iTope score) of the humanized variants as compared to parental. VH0 and humanized heavy (VH1-5) and light (VK1-5) chains used to generate humanized variants for lead drug selection. Heavy and light chains enabling combinations resulting in ⁇ 6 high affinity epitopes are in bold.
  • Example 8 Improved characteristics of the humanized anti-Nectin-4 mAbs (Improved developability)
  • Thermal ramp stability experiments are well established methods for ranking proteins and formulations for stability.
  • a protein’s denaturation profile provides information about its thermal stability and represents a structural ‘fingerprint’ for assessing structural and formulation buffer modifications.
  • a widely used measure of the thermal structural stability of a protein is the temperature at which it unfolds from the native state to a denatured state.
  • melting temperature For many proteins, this unfolding process occurs over a narrow temperature range and the mid-point of this transition is termed ‘melting temperature’ or ‘Tm’.
  • Tm melting temperature
  • UNcle measures the fluorescence of Sypro Orange (which binds to exposed hydrophobic regions of proteins) as the protein undergoes conformational changes.
  • Purified lead antibodies in duplicate, were diluted to a final test concentration of 0.5 mg/ml in PBS and into which Sypro Orange (at 160x Stock solution) was added to a final concentration of 20x solution. 9 pL of each sample mixture was loaded in duplicate into Uni microcuvettes.
  • Example 9 Improved characteristics of the humanized anti-Nectin-4 mAbs (Improved productivity)
  • the production of the mAb is a complex process, impacted by the amino acid composition and the structure of the molecule. Transient production is a good indicator of the “productivity” the mAb possess.
  • Chimeric (VHO/VKO) and combinations of humanized heavy and light chain DNA constructs were transiently transfected into HEK293 EBNA adherent cells (LGC Standards, Teddington, UK) using a PEI transfection method in 6 well plates and incubated for 6 days post-transfection. Samples were harvested and antibody concentrations were measured on the Octet QK 384 using Protein A biosensors (Molecular Devices, Wokingham, Berkshire, UK), using an IgGl antibody as standard.
  • the humanization process generally improved the productivity of all mAb (calculated by dividing the titer (mg/ml) of the relevant humanized variant to that of the parental/chimeric VHO/VKO), with >10-folds improved productivity of the leading clones (VH4/VK5 and VH5/VK5) as shown in Figure 5.
  • Enfortumab-vedotin is the only approved ADC targeting Nectin-4. Although having excellent clinical results, PADCEV has severe adverse events, some of which can be attributed to on-target/off-tumor binding, when PADCEVis able to act even at extremely low density of Nectin-4 which is found in some normal tissues. It is here hypothesized, that some of these adverse events can be prevented by an ADC with significantly lower on-target binding/activity.
  • Binding was evaluated on MDA-MB-468 cells. Briefly, 5*10 4 cells per well were stained using either PADCEV or NTX1105 conjugated to MMAE at the same DAR as PADCEV. The ADCs were added at concentrations starting 3 ug/ml using 3-fold dilutions. The detection was done using Alexa Fluor® 647 (AffiniPure Fab Fragment Goat Anti-Human IgG (H+L) (cat 109- 607-003 Jackson ImmunoResearch).
  • Example 12 Superior anti-tumor activity in-vivo of NTX1105 lead humanized clone compared to Enfortumab-vedotin (PADCEV).
  • PADCEV Enfortumab-vedotin
  • NTX1105 lead humanized clone VH4/VK5
  • PADCEV enfortumab-vedotin
  • Enfortumab-vedotin is the leading anti-Nectin-4 ADC, already approved for the treatment of patients with urothelial cancer.
  • the lead ADC humanized NTX1105 (VH4/VK5), anti-Nectin-4 ADC
  • the tumor cells (5M cells/mouse) were implanted s.c. to 6 weeks old Nude female mice.
  • NTX1105 ADC was significantly superior to PADCEV in preventing tumor growth during the entire duration of the study.
  • PADCEV treated group no longer was significantly superior in terms of tumor volume compared to PBS.
  • NTX1 105 ADC treated group led to complete growth arrest of the tumor and had lower tumor volume compared to study start even at day 48 (110-day-48 compared to 138mm 3 ).
  • these results are unexpected, especially given the significantly stronger in-vitro activity of PADCEV.
  • this is the first time in which Nectin-4 targeting ADC (NTX1105) exhibited significant anti-tumor activity in a model in which no biological activity was seen for PADCEV.
  • Example 13 Humanized NTX1105(H4K5-FcgR null ) ADC can regress H322 tumors in vivo with either Tubulin or TOPO1 targeting payloads.
  • both ADCs were capable of regressing established tumors to the same extent, a surprising outcome considering previous attempts to use TOPO1 targeting payload (Figure 4). This shows the potential utility of NTX1105 ADC using both MMAE and TOPO1 payloads.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Cell Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Urology & Nephrology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne des anticorps humanisés dirigés contre la nectine-4 et des conjugués anticorps-médicament (ADC) des anticorps humanisés, ainsi qu'une utilisation associée dans le traitement de maladies, en particulier de cancer.
PCT/IL2023/051016 2022-09-20 2023-09-19 Anticorps humanisés dirigés contre la nectine-4 et conjugués médicamenteux associés WO2024062476A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263408106P 2022-09-20 2022-09-20
US63/408,106 2022-09-20

Publications (1)

Publication Number Publication Date
WO2024062476A1 true WO2024062476A1 (fr) 2024-03-28

Family

ID=88412466

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2023/051016 WO2024062476A1 (fr) 2022-09-20 2023-09-19 Anticorps humanisés dirigés contre la nectine-4 et conjugués médicamenteux associés

Country Status (1)

Country Link
WO (1) WO2024062476A1 (fr)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017042210A1 (fr) 2015-09-09 2017-03-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps présentant une spécificité pour la nectine-4 et leurs utilisations
WO2018158398A1 (fr) 2017-03-02 2018-09-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps présentant une spécificité pour la nectine-4 et leurs utilisations
WO2019215728A1 (fr) 2018-05-09 2019-11-14 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Anticorps spécifiques de la nectine-4 humaine
WO2021069508A1 (fr) * 2019-10-07 2021-04-15 Université D'aix-Marseille Anticorps ayant une spécificité pour la nectine -4 et leurs utilisations
WO2021213434A1 (fr) * 2020-04-21 2021-10-28 迈威(上海)生物科技股份有限公司 Anticorps contre la nectine-4 et son application
WO2022051591A2 (fr) * 2020-09-04 2022-03-10 Novarock Biotherapeutics, Ltd. Anticorps anti-nectine-4 et leurs utilisations
WO2022056304A1 (fr) * 2020-09-10 2022-03-17 TCR2 Therapeutics Inc. Compositions et méthodes pour la reprogrammation de tcr au moyen de protéines de fusion spécifiques de la nectine-4

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017042210A1 (fr) 2015-09-09 2017-03-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps présentant une spécificité pour la nectine-4 et leurs utilisations
WO2018158398A1 (fr) 2017-03-02 2018-09-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps présentant une spécificité pour la nectine-4 et leurs utilisations
WO2019215728A1 (fr) 2018-05-09 2019-11-14 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Anticorps spécifiques de la nectine-4 humaine
WO2021069508A1 (fr) * 2019-10-07 2021-04-15 Université D'aix-Marseille Anticorps ayant une spécificité pour la nectine -4 et leurs utilisations
WO2021213434A1 (fr) * 2020-04-21 2021-10-28 迈威(上海)生物科技股份有限公司 Anticorps contre la nectine-4 et son application
WO2022051591A2 (fr) * 2020-09-04 2022-03-10 Novarock Biotherapeutics, Ltd. Anticorps anti-nectine-4 et leurs utilisations
WO2022056304A1 (fr) * 2020-09-10 2022-03-17 TCR2 Therapeutics Inc. Compositions et méthodes pour la reprogrammation de tcr au moyen de protéines de fusion spécifiques de la nectine-4

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
"GenBank", Database accession no. Q96NY8-NECT4_HUMAN
CHAILTA-EID, CANCER RES., vol. 76, 2016, pages 3003 - 13
LEFRANC M-P ET AL., DEV COMP IMMUNOL, vol. 27, 2003, pages 55 - 77
PARK ET AL., MOLECULES, vol. 24, 2019, pages 2754
WU T.TKABAT E.A., J EXP MED, vol. 132, 1970, pages 211 - 50

Similar Documents

Publication Publication Date Title
RU2758234C2 (ru) ЛЕЧЕНИЕ ТРИЖДЫ НЕГАТИВНОГО РАКА МОЛОЧНОЙ ЖЕЛЕЗЫ, ХАРАКТЕРИЗУЮЩЕГОСЯ ЭКСПРЕССИЕЙ Trop-2, С ПОМОЩЬЮ САЦИТУЗУМАБА ГОВИТЕКАНА И ИНГИБИТОРА Rad51
US20210128742A1 (en) CD123 Antibodies and Conjugates Thereof
US10744129B2 (en) Therapy of small-cell lung cancer (SCLC) with a topoisomerase-I inhibiting antibody-drug conjugate (ADC) targeting Trop-2
US10799597B2 (en) Subcutaneous administration of antibody-drug conjugates for cancer therapy
CN108472361B (zh) 抗cd37免疫缀合物及抗cd20抗体组合物
CN115969970A (zh) Abcg2抑制剂与sacituzumab govitecan的组合
JP2019519481A (ja) チェックポイント阻害薬に再発/耐性を示す腫瘍を治療するための抗Trop−2−SN−38抗体薬物複合体の効果
EP3130356A1 (fr) Conjugués anticorps-médicament
TW201834697A (zh) Her2標靶抗體-藥物結合物之組合療法
JP2023113921A (ja) 抗folr1イムノコンジュゲートと抗pd-1抗体の組み合わせ
US20200283540A1 (en) Humanized anti-liv1 antibodies for the treatment of breast cancer
JP7188837B2 (ja) Trop-2を標的化するトポイソメラーゼ-i阻害抗体-薬物コンジュゲート(adc)による小細胞肺癌(sclc)の療法
WO2024062476A1 (fr) Anticorps humanisés dirigés contre la nectine-4 et conjugués médicamenteux associés
JP2023506158A (ja) Cd276に特異的な抗体-薬物コンジュゲートおよびその使用
WO2023161943A1 (fr) Anticorps humanisés dirigés contre la nectine-2 et conjugués médicamenteux de ceux-ci
WO2024047654A1 (fr) Conjugués médicamenteux d'anticorps anti-pvr humanisés
US20230330241A1 (en) Humanized anti-liv1 antibodies for the treatment of cancer
US20240158484A1 (en) Humanized anti-liv1 antibodies for the treatment of cancer
JP2024521667A (ja) 抗-cd205抗体及び免疫チェックポイント・インヒビターを含む組み合わせ医薬
WO2023228095A1 (fr) Schéma posologique d'un conjugué anticorps anti-cdh6-médicament
WO2024050031A2 (fr) Nouveaux dérivés de camptothécine en tant que charges utiles de conjugués anticorps-médicament (cam)
JP2024507275A (ja) 細胞間接着分子1(icam1)抗体薬物抱合体およびそれらの使用

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23789791

Country of ref document: EP

Kind code of ref document: A1